Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stuart Mair is active.

Publication


Featured researches published by Stuart Mair.


Xenobiotica | 2014

Metabolism and disposition of the dipeptidyl peptidase IV inhibitor teneligliptin in humans

Yoshinobu Nakamaru; Yoshiharu Hayashi; Ruriko Ikegawa; Shuji Kinoshita; Begonya Perez Madera; Dave Gunput; Atsuhiro Kawaguchi; Martin Davies; Stuart Mair; Hiroshi Yamazaki; Toshiyuki Kume; Masayuki Suzuki

Abstract 1. The absorption, metabolism and excretion of teneligliptin were investigated in healthy male subjects after a single oral dose of 20 mg [14C]teneligliptin. 2. Total plasma radioactivity reached the peak concentration at 1.33 h after administration and thereafter disappeared in a biphasic manner. By 216 h after administration, ≥90% of the administered radioactivity was excreted, and the cumulative excretion in the urine and faeces was 45.4% and 46.5%, respectively. 3. The most abundant metabolite in plasma was a thiazolidine-1-oxide derivative (designated as M1), which accounted for 14.7% of the plasma AUC (area under the plasma concentration versus time curve) of the total radioactivity. The major components excreted in urine were teneligliptin and M1, accounting for 14.8% and 17.7% of the dose, respectively, by 120 h, whereas in faeces, teneligliptin was the major component (26.1% of the dose), followed by M1 (4.0%). 4. CYP3A4 and FMO3 are the major enzymes responsible for the metabolism of teneligliptin in humans. 5. This study indicates the involvement of renal excretion and multiple metabolic pathways in the elimination of teneligliptin from the human body. Teneligliptin is unlikely to cause conspicuous drug interactions or changes in its pharmacokinetics patients with renal or hepatic impairment, due to a balance in the elimination pathways.


Drug Metabolism and Disposition | 2014

Assessment of the Mass Balance Recovery and Metabolite Profile of Avibactam in Humans and In Vitro Drug-Drug Interaction Potential

Karthick Vishwanathan; Stuart Mair; Anshul Gupta; James Atherton; Jacqueline Clarkson-Jones; Timi Edeki; Shampa Das

Avibactam, a novel non-β-lactam β-lactamase inhibitor with activity against Ambler class A, class C, and some class D enzymes is being evaluated in combination with various β-lactam antibiotics to treat serious bacterial infections. The in vivo mass balance recovery and metabolite profile of [14C] avibactam (500 mg/1-h infusion) was assessed in six healthy male subjects, and a series of in vitro experiments evaluated the metabolism and drug-drug interaction potential of avibactam. In the mass balance study, measurement of plasma avibactam (using a validated liquid chromatography-tandem mass spectrometry method) and total radioactivity in plasma, whole blood, urine, and feces (using liquid scintillation counting) indicated that most of the avibactam was excreted unchanged in urine within 12 hours, with recovery complete (>97% of the administered dose) within 96 hours. Geometric mean avibactam renal clearance (158 ml/min) was greater than the product of unbound fraction of drug and glomerular filtration rate (109.5 ml/min), suggesting that active tubular secretion accounted for some renal elimination. There was no evidence of metabolism in plasma and urine, with unchanged avibactam the major component in both matrices. Avibactam demonstrated in vitro substrate potential for organic anion transporters 1 and 3 (OAT1 and OAT3) proteins expressed in human embryonic kidney 293 cells (Km > 1000 μM; >10-fold the Cmax of a therapeutic dose), which could account for the active tubular secretion observed in vivo. Avibactam uptake by OAT1 and OAT3 was inhibited by probenecid, a potent OAT1/OAT3 inhibitor. Avibactam did not interact with various other membrane transport proteins or cytochrome P450 enzymes in vitro, suggesting it has limited propensity for drug–drug interactions involving cytochrome P450 enzymes.


The Journal of Clinical Pharmacology | 2014

Netupitant PET imaging and ADME studies in humans

Tulla Spinelli; Selma Calcagnile; Claudio Giuliano; Giorgia Rossi; Corinna Lanzarotti; Stuart Mair; Lloyd Stevens; Ian Nisbet

Netupitant is a new, selective NK1 receptor antagonist under development for the prevention of chemotherapy‐induced nausea and vomiting. Two studies were conducted to evaluate the brain receptor occupancy (RO) and disposition (ADME) of netupitant in humans. Positron emission tomography (PET) imaging with the NK1 receptor‐binding–selective tracer [11C]‐GR205171 was used to evaluate the brain penetration of different doses of netupitant (100, 300, and 450 mg) and to determine the NK1‐RO duration. A NK1‐RO of 90% or higher was achieved with all doses in the majority of the tested brain regions at Cmax, with a long duration of RO. The netupitant minimal plasma concentration predicted to achieve a NK1‐RO of 90%, C90%, in the striatum was 225 ng/mL; after administration of netupitant 300 mg, concentrations exceeded the C90%. In the ADME study, a single nominal dose of [14C]‐netupitant 300 mg was used to assess its disposition. Absorption was rapid and netupitant was extensively metabolized via Phase I and II hepatic metabolism. Elimination of >90% was predicted at day 29 and was principally via hepatic/biliary route (>85%) with a minor contribution of the renal route (<5%). In conclusion, these studies demonstrate that netupitant is a potent agent targeting NK1 receptors with long lasting RO. In addition, netupitant is extensively metabolized and is mainly eliminated through the hepatic/biliary route and to a lesser extent via the kidneys.


Xenobiotica | 2015

Disposition, metabolism and mass balance of delafloxacin in healthy human volunteers following intravenous administration.

Andrew McEwen; Laura Lawrence; Randy Hoover; Lloyd Stevens; Stuart Mair; Gill Ford; Dylan Williams; Stuart Wood

Abstract 1. The pharmacokinetics and disposition of delafloxacin was investigated following a single intravenous (300 mg, 100 µCi) dose to healthy male subjects. 2. Mean Cmax, AUC0–∞, Tmax and t1/2 values for delafloxacin were 8.98 µg/mL, 21.31 µg h/mL, 1 h and 2.35 h, respectively, after intravenous dosing. 3. Radioactivity was predominantly excreted via the kidney with 66% of the radioactive dose recovered in the urine. Approximately 29% of the radioactivity was recovered in the faeces, giving an overall mean recovery of 94% administered radioactivity. 4. The predominant circulating components were identified as delafloxacin and a direct glucuronide conjugate of delafloxacin.


Xenobiotica | 2014

Absorption, metabolism and excretion of [14C]gemigliptin, a novel dipeptidyl peptidase 4 inhibitor, in humans.

Namtae Kim; Lorna Patrick; Stuart Mair; Lloyd Stevens; Gill Ford; Vicky Birks; Sung-Hack Lee

Abstract 1. Gemigliptin (formerly known as LC15-0444) is a newly developed dipeptidyl peptidase 4 inhibitor for the treatment of type 2 diabetes. Following oral administration of 50 mg (5.4 MBq) [14C]gemigliptin to healthy male subjects, absorption, metabolism and excretion were investigated. 2. A total of 90.5% of administered dose was recovered over 192 hr postdose, with 63.4% from urine and 27.1% from feces. Based on urinary recovery of radioactivity, a minimum 63.4% absorption from gastrointestinal tract could be confirmed. 3. Twenty-three metabolites were identified in plasma, urine and feces. In plasma, gemigliptin was the most abundant component accounting for 67.2% ∼ 100% of plasma radioactivity. LC15-0636, a hydroxylated metabolite of gemigliptin, was the only human metabolite with systemic exposure more than 10% of total drug-related exposure. Unchanged gemigliptin accounted for 44.8% ∼ 67.2% of urinary radioactivity and 27.7% ∼ 51.8% of fecal radioactivity. The elimination of gemigliptin was balanced between metabolism and excretion through urine and feces. CYP3A4 was identified as the dominant CYP isozyme converting gemigliptin to LC15-0636 in recombinant CYP/FMO enzymes.


Xenobiotica | 2018

Absorption, Disposition and Metabolic Pathway of Amiselimod (MT-1303) in Healthy Volunteers in a Mass Balance Study

Takayuki Kifuji; Shinsuke Inoue; Megumi Furukawa; Begonya Perez Madera; Takahiro Goto; Hiroshi Kumagai; Stuart Mair; Atsuhiro Kawaguchi

Abstract The absorption, metabolism and excretion of MT-1303 were investigated in healthy male subjects after a single oral dose of 0.4 mg [14C]-MT-1303 (ClinicalTrials.gov NCT02293967). The MT-1303 concentration in the plasma reached a maximum at 12 h after administration. Thereafter, the concentration declined with a half-life of 451 h. At the final assessment on Day 57, 91.16% of the administered radioactivity was excreted, and the cumulative excretion in the urine and faeces was 35.32% and 55.84%, respectively. The most abundant metabolite in plasma was MT-1303-P, which accounted for 42.6% of the area under the plasma concentration–time curve (AUC) of the total radioactivity. The major component excreted in urine was Human Urine (HU)4 (3066434), accounting for 28.1% of radioactivity in the sample (4.05% of the dose), whereas MT-1303 was a major component in the faeces, accounting for 89.8% of radioactivity in the sample (25.49% of the dose) up to 240 h after administration. This study indicates that multiple metabolic pathways are involved in the elimination of MT-1303 from the human body and the excretion of MT-1303 and MT-1303-P via the kidney is low. Therefore, MT-1303 is unlikely to cause conspicuous drug interactions or alter pharmacokinetics in patients with renal impairment. Trial registration: ClinicalTrials.gov identifier: NCT02293967.


European Respiratory Journal | 2012

Design and implementation of a multi-part, flexible protocol to assess the tolerability and pharmacodynamic effects of PUR118 in healthy subjects and COPD patients

Lorna Patrick; Alyson Connor; Stuart Mair; Joanne Collier; Molly Rosano; John W. Hanrahan


Cancer Chemotherapy and Pharmacology | 2017

Novel formulation of abiraterone acetate might allow significant dose reduction and eliminates substantial positive food effect

Tamás Solymosi; Zsolt Ötvös; Réka Angi; Betti Ordasi; Tamás Jordán; László Molnár; John McDermott; Vanessa Zann; Ann Church; Stuart Mair; Genovéva Filipcsei; Gábor Heltovics; Hristos Glavinas


Journal of Clinical Oncology | 2018

Novel formulation of abiraterone acetate with significantly higher bioavailability and eliminated food effect might allow higher abiraterone exposure and better patient outcome.

Hristos Glavinas; Tamás Solymosi; Zsolt Ötvös; Réka Angi; Betti Ordasi; Tamás Jordán; László Molnár; John McDermott; Vanessa Zann; Ann Church; Stuart Mair; Genovéva Filipcsei; Gábor Heltovics


Drug Metabolism and Pharmacokinetics | 2018

An open-label, two-period study designed to evaluate and compare the mass balance recovery and metabolite profile of 14 c deutetrabenazine and 14 c-tetrabenazine in healthy male subjects

Margaret Bradbury; David Stamler; Thomas A. Baillie; Stuart Mair; Sharan Sidhu; Stuart Wood; Iain Shaw

Collaboration


Dive into the Stuart Mair's collaboration.

Top Co-Authors

Avatar

Lloyd Stevens

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Ann Church

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Iain Shaw

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

John McDermott

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Lorna Patrick

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Vanessa Zann

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alyson Connor

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Helen Walker

University of Nottingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge